Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cancer Cell ; 11(3): 275-89, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17349584

RESUMO

The FEZ1/LZTS1 (LZTS1) protein is frequently downregulated in human cancers of different histotypes. LZTS1 is expressed in normal tissues, and its introduction in cancer cells inhibits cell growth and suppresses tumorigenicity, owing to an accumulation of cells in G2/M. Here, we define its role in cell cycle regulation and tumor progression by generating Lzts1 knockout mice. In Lzts1(-/-) mouse embryo fibroblasts (MEFs), Cdc25C degradation was increased during M phase, resulting in decreased Cdk1 activity. As a consequence, Lzts1(-/-) MEFs showed accelerated mitotic progression, resistance to taxol- and nocodazole-induced M phase arrest, and improper chromosome segregation. Accordingly, Lzts1 deficiency was associated with an increased incidence of both spontaneous and carcinogen-induced cancers in mice.


Assuntos
Proteína Quinase CDC2/fisiologia , Proteínas de Ciclo Celular/fisiologia , Transformação Celular Neoplásica , Mitose , Neoplasias Gástricas/patologia , Proteínas Supressoras de Tumor/fisiologia , Fosfatases cdc25/fisiologia , Animais , Antineoplásicos/farmacologia , Carcinógenos , Divisão Celular , Células Cultivadas , Segregação de Cromossomos , Dimetilnitrosamina/análogos & derivados , Fibroblastos/metabolismo , Fibroblastos/patologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Nocodazol/farmacologia , Paclitaxel/farmacologia , Neoplasias Gástricas/induzido quimicamente , Neoplasias Gástricas/genética , Proteínas Supressoras de Tumor/genética
2.
Carcinogenesis ; 34(3): 570-6, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23222816

RESUMO

Upper aerodigestive tract (UADT) cancers of the oral cavity and esophagus are a significant global health burden, and there is an urgent need to develop relevant animal models to identify chemopreventive and therapeutic strategies to combat these diseases. Antizyme (AZ) is a multifunctional negative regulator of cellular polyamine levels, and here, we evaluate the susceptibility of keratin 5 (K5)-AZ transgenic mice to tumor models that combine chemical carcinogenesis with dietary and genetic risk factors known to influence human susceptibility to UADT cancer and promote UADT carcinogenesis in mice. First, p53(+/-) and K5-AZ/p53(+/-) (AZ/p53(+/-)) mice were placed on a zinc-deficient (ZD) or zinc-sufficient (ZS) diet and chronically exposed to 4-nitroquinoline 1-oxide. Tongue tumor incidence, multiplicity and size were substantially reduced in both ZD and ZS AZ/p53(+/-) mice compared with p53(+/-). AZ expression also reduced progression to carcinoma in situ or invasive carcinoma and decreased expression of the squamous cell carcinoma biomarkers K14, cyclooxygenase-2 and metallothionein. Next, AZ-expressing p53(+/-) and p53 null mice were placed on the ZD diet and treated with a single dose of N-nitrosomethylbenzylamine. Regardless of p53 status, forestomach (FST) tumor incidence, multiplicity and size were greatly reduced with AZ expression, which was also associated with a significant decrease in FST epithelial thickness along with reduced proliferation marker K6 and increased differentiation marker loricrin. These studies demonstrate the powerful tumor suppressive effects of targeted AZ expression in two distinct and unique mouse models and validate the polyamine metabolic pathway as a target for chemoprevention of UADT cancers.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Transformação Celular Neoplásica/metabolismo , Neoplasias Esofágicas/metabolismo , Proteínas/metabolismo , Neoplasias da Língua/metabolismo , 4-Nitroquinolina-1-Óxido , Animais , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/patologia , Proliferação de Células , Dimetilnitrosamina/análogos & derivados , Epitélio/patologia , Neoplasias Esofágicas/induzido quimicamente , Neoplasias Esofágicas/patologia , Mucosa Gástrica/patologia , Neoplasias de Cabeça e Pescoço/induzido quimicamente , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Poliaminas/metabolismo , Proteínas/genética , Quinolonas , Neoplasias da Língua/induzido quimicamente , Neoplasias da Língua/patologia , Carga Tumoral , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
3.
Carcinogenesis ; 33(9): 1736-44, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22689922

RESUMO

Zinc deficiency (ZD) increases the risk of esophageal squamous cell carcinoma (ESCC). In a rat model, chronic ZD induces an inflammatory gene signature that fuels ESCC development. microRNAs regulate gene expression and are aberrantly expressed in cancers. Here we investigated whether chronic ZD (23 weeks) also induces a protumorigenic microRNA signature. Using the nanoString technology, we evaluated microRNA profiles in ZD esophagus and six additional tissues (skin, lung, pancreas, liver, prostate and peripheral blood mononuclear cells [PBMC]). ZD caused overexpression of inflammation genes and altered microRNA expression across all tissues analyzed, predictive of disease development. Importantly, the inflammatory ZD esophagus had a distinct microRNA signature resembling human ESCC or tongue SCC miRNAomes with miR-31 and miR-21 as the top-up-regulated species. Circulating miR-31 was also the top-up-regulated species in PBMCs. In ZD esophagus and tongue, oncogenic miR-31 and miR-21 overexpression was accompanied by down-regulation of their respective tumor-suppressor targets PPP2R2A and PDCD4. Importantly, esophageal miR-31 and miR-21 levels were directly associated with the appearance of ESCC in ZD rats, as compared with their cancer-free Zn-sufficient or Zn-replenished counterparts. In situ hybridization analysis in rat and human tongue SCCs localized miR-31 to tumor cells and miR-21 to stromal cells. In regressing tongue SCCs from Zn-supplemented rats, miR-31 and miR-21 expression was concomitantly reduced, establishing their responsiveness to Zn therapy. A search for putative microRNA targets revealed a bias toward genes in inflammatory pathways. Our finding that ZD causes miR-31 and miR-21 dysregulation associated with inflammation provides insight into mechanisms whereby ZD promotes ESCC.


Assuntos
Carcinoma de Células Escamosas/etiologia , Neoplasias Esofágicas/etiologia , MicroRNAs/fisiologia , Zinco/deficiência , Animais , Proteínas Reguladoras de Apoptose/genética , Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Esôfago/metabolismo , Humanos , Masculino , MicroRNAs/análise , Proteínas de Ligação a RNA/genética , Ratos , Ratos Sprague-Dawley , Língua/metabolismo , Neoplasias da Língua/genética , Zinco/administração & dosagem
4.
Carcinogenesis ; 32(4): 554-60, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21245412

RESUMO

Dietary zinc (Zn) deficiency is implicated in the pathogenesis of human oral-esophageal cancers. In rats, Zn deficiency causes increased cell proliferation and cyclooxygenase-2 (COX-2) overexpression and enhances oral carcinogenesis by 4-nitroquinoline 1-oxide (NQO). Zn replenishment reverses all these effects. We questioned whether Zn has antitumor efficacy in a Zn-sufficient animal by investigating in Zn-sufficient rats (i) the efficacy of Zn supplementation on the progression of tongue squamous cell carcinogenesis induced by drinking water exposure to high (20-30 p.p.m.) and low (10 p.p.m.) doses of NQO and (ii) the modulating effects of Zn supplementation on biomarker expression in tongue lesions by immunohistochemistry. In rats exposed to high doses of NQO, Zn supplementation significantly reduced the incidence of papillomas from 100 to 64.7% (P=0.018) and invasive carcinomas from 93.8 to 52.9% (P=0.017). In rats exposed to low doses of NQO, where only minimally invasive carcinomas developed, Zn supplementation significantly reduced tumor multiplicity, incidence of tumors (1-2 mm), hyperplasia, dysplasia, papillomas and progression to carcinoma. Immunohistochemical analysis of carcinomas showed that Zn supplementation caused a shift to a less proliferative/aggressive cancer phenotype by reducing cell proliferation, stimulating apoptosis and decreasing expression of the key tumor markers cyclin D1, p53 and COX-2. Additionally, Zn supplementation significantly reduced cell proliferation in non-lesional tongue squamous epithelia, thereby suppressing tumor development. Together, the results demonstrate that Zn supplementation has chemopreventive efficacy against oral carcinogenesis in nutritionally complete animals. Our data suggest that Zn supplementation may be efficacious in the chemoprevention of human oral cancer.


Assuntos
4-Nitroquinolina-1-Óxido/toxicidade , Neoplasias da Língua/prevenção & controle , Zinco/administração & dosagem , Animais , Apoptose , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/prevenção & controle , Proliferação de Células , Suplementos Nutricionais , Masculino , Ratos , Ratos Sprague-Dawley , Neoplasias da Língua/induzido quimicamente , Neoplasias da Língua/patologia , Zinco/sangue
5.
Int J Cancer ; 129(2): 331-45, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20857495

RESUMO

Zinc (Zn)-deficiency (ZD) is implicated in the pathogenesis of human oral-esophageal cancers. Previously, we showed that in ZD mice genetic deletion of cyclooxygenase-2 (Cox-2) enhances N-nitrosomethylbenzylamine-induced forestomach carcinogenesis. By contrast, Cox-2 deletion offers protection in Zn-sufficient (ZS) mice. We hypothesize that ZD activates pathways insensitive to COX-2 inhibition, thereby promoting carcinogenesis. This hypothesis is tested in a Cox-2(-/-) mouse tongue cancer model that mimics pharmacologic blockade of COX-2 by firstly examining transcriptome profiles of forestomach mucosa from Cox-2(-/-) and wild-type mice on a ZD vs. ZS diet, and secondly investigating the roles of identified markers in mouse forestomach/tongue preneoplasia and carcinomas. In Cox-2(-/-) mice exposed to the tongue carcinogen 4-nitroquinoline 1-oxide, dietary ZD elicited tongue/esophagus/forestomach carcinomas that were prevented by ZS. The precancerous ZD:Cox-2(-/-) vs. ZS:Cox-2(-/-) forestomach had an inflammatory signature with upregulation of the proinflammation genes S100a8 and S100a9. Bioinformatics analysis revealed overrepresentation of inflammation processes comprising S100a8/a9 and an nuclear factor (NF)-κB network with connectivity to S100A8. Immunohistochemistry revealed co-overexpression of S100A8, its heterodimeric partner S100A9, the receptor for advanced glycation end-products (RAGE), NF-κB p65, and cyclin D1, in ZD:Cox-2(-/-) forestomach/tongue preneoplasia and carcinomas, evidence for the activation of a RAGE-S100A8/A9 inflammatory pathway. Accumulation of p53 in these carcinomas indicated activation of additional inflammatory pathways. Zn-replenishment in ZD:Cox-2(-/-) mice reversed the inflammation and inhibited carcinogenesis. Thus, ZD activates alternative inflammation-associated cancer pathways that fuel tumor progression and bypass the antitumor effect of Cox-2 ablation. These findings have important clinical implications, as combination cancer therapy that includes Zn may improve efficacy.


Assuntos
Calgranulina A/metabolismo , Ciclo-Oxigenase 2/deficiência , Neoplasias Gástricas/patologia , Neoplasias da Língua/patologia , Zinco/deficiência , 4-Nitroquinolina-1-Óxido , Animais , Carcinógenos , Ciclo-Oxigenase 2/genética , Dimetilnitrosamina/análogos & derivados , Modelos Animais de Doenças , Progressão da Doença , Neoplasias Esofágicas/induzido quimicamente , Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/patologia , Feminino , Deleção de Genes , Inflamação/enzimologia , Masculino , Camundongos , Camundongos Knockout , Neoplasias Gástricas/induzido quimicamente , Neoplasias Gástricas/enzimologia , Neoplasias da Língua/induzido quimicamente , Neoplasias da Língua/enzimologia
6.
Gastroenterology ; 136(3): 953-66, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19111725

RESUMO

BACKGROUND & AIMS: Zinc deficiency is implicated in the pathogenesis of human esophageal cancer. In the rat esophagus, it induces cell proliferation, modulates genetic expression, and enhances carcinogenesis. Zinc-replenishment reverses proliferation and inhibits carcinogenesis. The zinc-deficient rat model allows the identification of biological differences affected by zinc during early esophageal carcinogenesis. METHODS: We evaluated gene expression profiles of esophageal epithelia from zinc-deficient and replenished rats vs zinc-sufficient rats using microarray analysis. We characterized the role of the top-up-regulated gene S100A8 in esophageal hyperplasia/reversal and in chemically induced esophageal carcinogenesis in zinc-modulated animals by immunohistochemistry and real-time quantitative polymerase chain reaction. RESULTS: The hyperplastic-deficient esophagus has a distinct expression signature with the proinflammation genes S100 calcium binding protein A8 (S100A8) and A9 (S100A9) up-regulated 57-fold and 5-fold, respectively. Zinc replenishment rapidly restored to control levels the expression of S100A8/A9 and 27 other genes and reversed the hyperplastic phenotype. With its receptor for advanced glycation end products (RAGE), colocalization and overexpression of S100A8 protein occurred in the deficient esophagus that overexpressed nuclear factor kappaBeta p65 and cyclooxygenase-2 (COX-2) protein. Zinc replenishment, but not a COX-2 inhibitor, reduced the overexpression of these 4 proteins. Additionally, esophageal S100A8/A9 messenger RNA levels were associated directly with the diverse tumorigenic outcome in zinc-deficient and zinc-replenished rats. CONCLUSIONS: In vivo zinc regulates S100A8 expression and modulates the link between S100A8-RAGE interaction and downstream nuclear factor kappaBeta/COX-2 signaling. The finding that zinc regulates an inflammatory pathway in esophageal carcinogenesis may lead to prevention and therapy for this cancer.


Assuntos
Calgranulina A/genética , Neoplasias Esofágicas/prevenção & controle , Lesões Pré-Cancerosas/tratamento farmacológico , Zinco/deficiência , Zinco/farmacologia , Animais , Calgranulina A/metabolismo , Ciclo-Oxigenase 2/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Esôfago/patologia , Esôfago/fisiologia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Hiperplasia , Masculino , Fenótipo , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Ratos , Ratos Sprague-Dawley , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Transcrição RelA/metabolismo
7.
Cancer Res ; 67(12): 5606-10, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17575124

RESUMO

The WWOX gene encodes a tumor suppressor spanning the second most common human fragile site, FRA16D. Targeted deletion of the Wwox gene in mice led to an increased incidence of spontaneous and ethyl nitrosourea-induced tumors. In humans, loss of heterozygosity and reduced or loss of WWOX expression has been reported in esophageal squamous cell cancers (SCC). In the present study, we examined whether inactivation of the Wwox gene might lead to enhanced esophageal/forestomach tumorigenesis induced by N-nitrosomethylbenzylamine. Wwox+/- and Wwox+/+ mice were treated with six intragastric doses of N-nitrosomethylbenzylamine and observed for 15 subsequent weeks. Ninety-six percent (25 of 26) of Wwox+/- mice versus 29% (10 of 34) of Wwox+/+ mice developed forestomach tumors (P = 1.3 x 10(-7)). The number of tumors per forestomach was significantly greater in Wwox+/- than in Wwox+/+ mice (3.2 +/- 0.34 versus 0.47 +/- 0.17; P < 0.0001). In addition, 27% of Wwox+/- mice had invasive SCC in the forestomach, as compared with 0% of wild-type controls (P = 0.002). Intriguingly, forestomachs from Wwox+/- mice displayed moderately strong Wwox protein staining in the near-normal epithelium, but weak and diffuse staining in SCC in the same tissue section, a result suggesting that Wwox was haploinsufficient for the initiation of tumor development. Our findings provide the first in vivo evidence of the tumor suppressor function of WWOX in forestomach/esophageal carcinogenesis and suggest that inactivation of one allele of WWOX accelerates the predisposition of normal cells to malignant transformation.


Assuntos
Inativação Gênica , Predisposição Genética para Doença , Neoplasias de Células Escamosas/genética , Oxirredutases/genética , Neoplasias Gástricas/genética , Animais , Carcinógenos/toxicidade , Transformação Celular Neoplásica/genética , Dimetilnitrosamina/análogos & derivados , Dimetilnitrosamina/toxicidade , Progressão da Doença , Neoplasias Esofágicas/induzido quimicamente , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Imuno-Histoquímica , Camundongos , Neoplasias de Células Escamosas/induzido quimicamente , Neoplasias de Células Escamosas/patologia , Neoplasias Gástricas/induzido quimicamente , Neoplasias Gástricas/patologia , Oxidorredutase com Domínios WW
8.
Int J Cancer ; 122(5): 978-89, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17985342

RESUMO

Zinc deficiency in humans is associated with an increased risk of upper aerodigestive tract (UADT) cancer. In rodents, zinc deficiency predisposes to carcinogenesis by causing proliferation and alterations in gene expression. We examined whether in zinc-deficient rodents, targeted disruption of the cyclooxygenase (COX)-2 pathway by the COX-2 selective inhibitor celecoxib or by genetic deletion prevent UADT carcinogenesis. Tongue cancer prevention studies were conducted in zinc-deficient rats previously exposed to a tongue carcinogen by celecoxib treatment with or without zinc replenishment, or by zinc replenishment alone. The ability of genetic COX-2 deletion to protect against chemically-induced forestomach tumorigenesis was examined in mice on zinc-deficient versus zinc-sufficient diet. The expression of 3 predictive biomarkers COX-2, nuclear factor (NF)-kappa B p65 and leukotriene A(4) hydrolase (LTA(4)H) was examined by immunohistochemistry. In zinc-deficient rats, celecoxib without zinc replenishment reduced lingual tumor multiplicity but not progression to malignancy. Celecoxib with zinc replenishment or zinc replenishment alone significantly lowered lingual squamous cell carcinoma incidence, as well as tumor multiplicity. Celecoxib alone reduced overexpression of the 3 biomarkers in tumors slightly, compared with intervention with zinc replenishment. Instead of being protected, zinc-deficient COX-2 null mice developed significantly greater tumor multiplicity and forestomach carcinoma incidence than wild-type controls. Additionally, zinc-deficient COX-2-/- forestomachs displayed strong LTA(4)H immunostaining, indicating activation of an alternative pathway under zinc deficiency when the COX-2 pathway is blocked. Thus, targeting only the COX-2 pathway in zinc-deficient animals did not prevent UADT carcinogenesis. Our data suggest zinc supplementation should be more thoroughly explored in human prevention clinical trials for UADT cancer.


Assuntos
Carcinoma de Células Escamosas/prevenção & controle , Ciclo-Oxigenase 2/deficiência , Neoplasias de Cabeça e Pescoço/prevenção & controle , Zinco/deficiência , Animais , Carcinógenos/toxicidade , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/etiologia , Celecoxib , Ciclo-Oxigenase 2/efeitos dos fármacos , Ciclo-Oxigenase 2/genética , Inibidores de Ciclo-Oxigenase 2/farmacologia , Neoplasias Esofágicas/induzido quimicamente , Neoplasias Esofágicas/prevenção & controle , Neoplasias de Cabeça e Pescoço/induzido quimicamente , Neoplasias de Cabeça e Pescoço/etiologia , Imuno-Histoquímica , Leucotrieno A4/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Sulfonamidas/farmacologia , Neoplasias da Língua/induzido quimicamente , Neoplasias da Língua/prevenção & controle , Fator de Transcrição RelA/metabolismo
9.
Cancer Res ; 65(17): 7790-9, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16140947

RESUMO

Zinc deficiency in rats enhances esophageal cell proliferation, causes alteration in gene expression, and promotes esophageal carcinogenesis. Zinc replenishment rapidly induces apoptosis in the esophageal epithelium thereby reversing cell proliferation and carcinogenesis. To identify zinc-responsive genes responsible for these divergent effects, we did oligonucleotide array-based gene expression profiling analyses in the precancerous zinc-deficient esophagus and in zinc-replenished esophagi after treatment with intragastric zinc compared with zinc-sufficient esophagi. Thirty-three genes (21 up-regulated and 12 down-regulated) showed a > or = 2-fold change in expression in the hyperplastic zinc-deficient versus zinc-sufficient esophageal epithelia. Expression of genes involved in cell division, survival, adhesion, and tumorigenesis were markedly changed. The zinc-sensitive gene metallothionein-1 (MT-1 was up-regulated 7-fold, the opposite of results for small intestine and liver under zinc-deficient conditions. Keratin 14 (KRT14, a biomarker in esophageal tumorigenesis), carbonic anhydrase II (CAII, a regulator of acid-base homeostasis), and cyclin B were up-regulated >4-fold. Immunohistochemistry showed that metallothionein and keratin 14 proteins were overexpressed in zinc-deficient esophagus, as well as in lingual and esophageal squamous cell carcinoma from carcinogen-treated rats, emphasizing their roles in carcinogenesis. Calponin 1 (CNN1, an actin cross-linking regulator) was down-regulated 0.2-fold. Within hours after oral zinc treatment, the abnormal expression of 29 of 33 genes returned to near zinc-sufficient levels, accompanied by reversal of the precancerous phenotype. Thus, we have identified new molecular markers in precancerous esophagus and showed their restoration by zinc replenishment, providing insights into the interaction between zinc and gene expression in esophageal cancer development and prevention.


Assuntos
Neoplasias Esofágicas/etiologia , Regulação Neoplásica da Expressão Gênica , Lesões Pré-Cancerosas/etiologia , Zinco/deficiência , 4-Nitroquinolina-1-Óxido/farmacologia , Animais , Carcinoma de Células Escamosas/etiologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Imuno-Histoquímica , Queratina-14 , Queratinas/biossíntese , Queratinas/genética , Masculino , Metalotioneína/biossíntese , Metalotioneína/genética , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima , Zinco/administração & dosagem
10.
Cancer Res ; 63(14): 3945-54, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12873989

RESUMO

Antizyme (AZ) is known to be a regulator of polyamine metabolism that inhibits ornithine decarboxylase activity and polyamine transport, thus restricting polyamine levels. Transgenic mice with AZ expression targeted to the basal cell layer of the forestomach epithelium by the keratin 5 promoter were used to investigate whether AZ overexpression inhibited uncontrolled cell proliferation in zinc-deficient (ZD) mice and reduced their susceptibility to forestomach carcinogenesis by N-nitrosomethylbenzylamine (NMBA). Four-week-old keratin 5/AZ and wild-type (Wt) littermates were placed on ZD or zinc-sufficient (ZS) diets to form four groups: ZD:AZ, ZD:Wt, ZS:AZ, and ZS:Wt. After 5 weeks, 27-45 mice in each group were treated twice with NMBA and sacrificed 14 weeks later. Independent of zinc intake, AZ mice had significantly lower forestomach tumor incidence and tumor multiplicity than respective Wt littermates (P < 0.001): 21% of ZD:AZ versus 76% of ZD:Wt mice and 3% of ZS:AZ versus 33% of ZS:Wt mice developed tumors. Spermidine content was reduced in NMBA-treated ZD:AZ forestomachs. Zinc deficiency increased the forestomach cell proliferation in Wt mice, but this effect was blocked by AZ. Conversely, apoptosis was substantially higher in control and NMBA-treated ZD:AZ than respective ZD:Wt forestomachs. The restored ZD:AZ forestomach epithelium displayed strong expression of Bax, a proapoptotic protein, and weak staining of cyclin D1 and its catalytic partner Cdk4, key regulatory proteins controlling G(1) to S progression. In contrast, proliferative ZD:Wt forestomach showed strong expression of Bcl-2, an antiapoptotic protein, and overexpression of cyclin D1/Cdk4. Treatment of ZD:Wt mice with alpha-difluoromethylornithine, an inhibitor of ornithine decarboxylase, had similar results to AZ in reducing tumor incidence, spermidine content, decreasing cell proliferation, and increasing apoptosis. These results demonstrate that AZ may act as a tumor suppressor gene stimulating apoptosis and restraining cell proliferation, thereby inhibiting forestomach tumor development. Although effects of AZ on functions other than polyamine metabolism are possible, alterations in polyamines are the most likely explanation for the reduction in tumors, supporting the use of strategies to modulate polyamine levels for cancer chemoprevention in individuals at high risk of developing malignancies of the gastrointestinal tract.


Assuntos
Apoptose/fisiologia , Dimetilnitrosamina/análogos & derivados , Biossíntese de Proteínas , Neoplasias Gástricas/prevenção & controle , Animais , Poliaminas Biogênicas/metabolismo , Carcinógenos , Divisão Celular/fisiologia , Eflornitina/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas/genética , Neoplasias Gástricas/induzido quimicamente , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Zinco/deficiência
11.
Cancer Res ; 63(14): 4244-52, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12874033

RESUMO

Overexpression of cyclin D1 and disruption of cell cycle control in G(1) occur frequently in human esophageal cancer. Transgenic (TG) mice with cyclin D1 overexpression targeted to the oral-esophageal tissue by the EBV ED-L2 promoter showed increased severity in esophageal dysplasia without cancer development, after multiple doses of N-nitrosomethylbenzylamine (NMBA). Dietary zinc deficiency (ZD) in mice enhances cellular proliferation in esophagus/forestomach and susceptibility to NMBA-induced carcinogenesis. We investigated whether cyclin D1 overexpression in TG mice, together with ZD, might lead to unchecked cell proliferation and accelerated NMBA-induced tumorigenesis. Five-week-old TG and wild-type (WT) mice were fed a ZD- or -sufficient (ZS) diet, forming four groups: ZD:TG; ZS:TG; ZD:WT; and ZS:WT. After 4 weeks, animals were given a single intragastric NMBA dose and were sacrificed 25 and 77 days later. Without NMBA, cell proliferation was greatest in ZD:TG esophagus/forestomach, followed by ZD:WT, and then ZS:TG>/=ZS:WT. The high rate of cell proliferation was accompanied by overexpression of cell cycle progression and tumorigenesis biomarkers, including proliferating cell nuclear antigen, cyclin D1, cyclin-dependent kinase 4, p53, cytokeratin 14, epidermal growth factor receptor, and by a reduced rate of apoptosis. ZD substantially increased forestomach tumor incidence in TG mice: 85% of ZD:TG versus 14% of ZS:TG mice had forestomach tumors (P < 0.001), with progression to malignancy occurring only in ZD:TG tumors. Additionally, 14% of ZD:TG mice developed esophageal tumors and esophageal intestinal metaplasia at 77 days. Thus, cyclin D1 overexpression, in cooperation with ZD, decontrols cell proliferation, ensuring cell expansion, a prerequisite for cancer development.


Assuntos
Ciclina D1/biossíntese , Neoplasias Esofágicas/metabolismo , Neoplasias Gástricas/metabolismo , Zinco/deficiência , Animais , Apoptose/fisiologia , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Ciclina D1/genética , Progressão da Doença , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia
12.
Cancer Res ; 63(1): 186-95, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12517797

RESUMO

The p53 tumor suppressor protein plays a pivotal role in preventing uncontrolled cellular proliferation. By contrast, zinc deprivation enhances esophageal cell proliferation and the induction of esophageal tumors in rodents by N-nitrosomethylbenzylamine (NMBA). We investigated whether p53 deficiency rendered zinc-deficient (ZD) mice more susceptible to NMBA-induced esophageal/forestomach carcinogenesis. At 6-7 weeks of age, p53 null (-/-), heterozygous (+/-), and wild-type (+/+) mice were placed on ZD or zinc-sufficient (ZS) diets to form six experimental groups: ZD:p53-/-; ZD:p53+/-; ZD:p53+/+; ZS:p53-/-; ZS:p53+/-; and ZS:p53+/+. After 3 weeks, 15-23 mice in each group were treated once with NMBA (2 mg/kg body weight). Control animals were untreated. Zinc deficiency alone induced unrestrained cellular proliferation in the esophagus and forestomach of p53-/- mice. Forestomach tumors were first detected in a ZD:p53-/- mouse at 13 days. By 30 days, 100% (21 of 21) of ZD:p53-/- mice developed forestomach tumors and 38% showed esophageal tumors versus 42 and 0% in ZS:p53-/- mice (P < 0.004, esophagus; P < 0.001, forestomach). ZD:p53-/- mice showed an accelerated progression to malignancy, with 10% of esophageal tumors and 38% of forestomach tumors presenting as carcinomas. Nearly 20% of ZD:p53-/- mice developed esophageal Barrett's metaplasia, a lesion not previously seen in NMBA-induced neoplasia. ZD:p53+/- mice had significantly higher tumor incidence than ZS:p53+/- mice. The order of tumor incidence in forestomach was as follows: naught incidence in ZS:p53+/+ mice; ZD:p53-/- > ZD:p53+/- > ZS:p53-/- > ZD:p53+/+ >/= ZS:p53+/- > ZS:p53+/+. The rapid rate of tumor induction/progression in ZD:p53-/- mice was accompanied by an increase in the rate of cell proliferation and a decrease in apoptosis. cDNA array expression analysis of known genes identified a 5-fold up-regulation of cytokeratin 14 mRNA expression in ZD:p53-/- versus ZS:p53-/- forestomach, a result showing gene-modulating effects of zinc deficiency. Cytokeratin 14 is a biomarker in human esophageal carcinogenesis. Our findings provide in vivo evidence for the collaboration of a deficiency of both p53 and zinc in esophageal carcinogenesis and reveal molecular targets of this collaboration.


Assuntos
Dimetilnitrosamina/análogos & derivados , Neoplasias Esofágicas/patologia , Neoplasias Gástricas/patologia , Proteína Supressora de Tumor p53/deficiência , Zinco/deficiência , Animais , Carcinógenos , Divisão Celular , Cruzamentos Genéticos , Dieta , Progressão da Doença , Neoplasias Esofágicas/induzido quimicamente , Neoplasias Esofágicas/genética , Genótipo , Queratinas/análise , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Neoplasias Gástricas/induzido quimicamente , Neoplasias Gástricas/genética , Proteína Supressora de Tumor p53/genética
13.
Oncogene ; 23(49): 8040-8, 2004 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-15361827

RESUMO

Zinc deficiency (ZD) in rats increases esophageal cell proliferation and the incidence of N-nitrosomethylbenzylamine-induced esophageal tumors. Conversely, zinc replenishment (ZR) rapidly induces apoptosis in esophageal epithelia and reverses cancer development. We investigated gene expression changes in ZR versus ZD esophageal epithelia to identify differentially expressed genes associated with the antitumor effect of ZR. Weanling rats were fed a ZD diet for 6 weeks to establish esophageal cell proliferation or a zinc-sufficient (ZS) diet. Then, 10 ZD rats were treated with zinc gluconate intragastrically and switched to ZS diet; the remaining 10 ZD and ZS animals were treated with saline. All animals were killed 26-28 h later. Using cDNA microarrays, real-time polymerase chain reaction amplification and RNA hybridization techniques, we identified novel differentially expressed genes, including a RNA-binding protein with two RNA recognition motifs and a zinc knuckle (ZD7), and a DNA/RNA helicase with a DEAD box (ZD10) with two splice variants, ZD10a and ZD10b. In situ hybridization detected increased mRNA expression of ZD7, ZD10a and ZD10b in ZR esophageal epithelia, which displayed markedly increased occurrence of apoptotic cells, relative to ZD epithelia. Overexpression of ZD7 in human esophageal cancer cells resulted in induction of apoptosis and activation of caspase-3 and -7, activities that were inhibited by caspase-specific inhibitors. In addition, ZD7 mRNA levels and zinc-induced apoptosis in rat squamous carcinoma cells were reduced by specific small interfering ribonucleic acids. Thus, ZR rapidly induces ZD7 and ZD10 expression, which in turn stimulates apoptosis. These results provide the beginnings of a molecular pathway for zinc-induced apoptosis under conditions that reverse esophageal tumor initiation.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Esofágicas/patologia , Perfilação da Expressão Gênica , Lesões Pré-Cancerosas/patologia , Zinco/farmacologia , Animais , Linhagem Celular Tumoral , Clonagem Molecular , Neoplasias Esofágicas/metabolismo , Masculino , Lesões Pré-Cancerosas/metabolismo , Ratos , Ratos Sprague-Dawley , Zinco/sangue , Zinco/deficiência
14.
Oncogene ; 23(22): 3990-8, 2004 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-15007387

RESUMO

To determine if: (1) 5' CpG island methylation is related to Fhit inactivation; (2) there are tumor or carcinogen-specific methylation patterns, we examined 35 CpG sites in the promoter, exon and intron 1 of the mouse Fhit gene. In primary tumors of lung, urinary bladder and tongue, induced by different carcinogens, 15-35% of sites were methylated, with specific methylation patterns associated with each cancer type, suggesting cancer- or tissue-specific methylation patterns. The methylation patterns were associated with reduced Fhit expression, as determined by immunohistochemical analyses. Methylation of rat Fhit 5' CpGs in mammary adenocarcinomas, detected by methylation specific PCR amplification, also correlated with reduced gene expression. Thus, there was an overall association between promoter/exon 1 methylation and decreased Fhit expression. In contrast, in cancer-derived cell lines 70-95% of the CpG sites were methylated. This is the first detailed study of the relationship between Fhit 5' CpG island methylation and Fhit expression in murine tumors, our main models for preclinical cancer studies, and provides evidence that loss of Fhit expression and methylation are correlated in these mouse models and these models will be useful to examine the complex relationships among gene expression, methylation patterns and organ specificity.


Assuntos
Hidrolases Anidrido Ácido/metabolismo , Expressão Gênica/fisiologia , Genes Reguladores , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Animais , Metilação de DNA , Humanos , Imuno-Histoquímica , Camundongos , Neoplasias/genética , Neoplasias/patologia
15.
FASEB J ; 17(12): 1768-70, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12958204

RESUMO

Fhit expression is reduced in most cancers, and Fhit replacement by FHIT expression viruses in lung, esophageal, pancreatic, and cervical cancers induces apoptosis in the cancer cells. Mice carrying one or two inactivated Fhit alleles are hypersensitive to development of N-nitrosomethylbenzylamine (NMBA)-induced forestomach tumors. In the present study, we investigated the kinetics and mechanism of tumor reversal and intervention by oral delivery of FHIT expression viruses. Tumor analysis showed that: a) by 37 days post-NMBA, control mice showed approximately 7 tumors and by 84 days approximately 10 tumors/forestomach; b) mice receiving FHIT virus at 2 or 42 days post-NMBA showed significantly reduced tumor burdens; c) Fhit was still expressed at 82 days postinfection; d) control viral infection had no effect on tumor development; and e) reduced Bcl2, increased Bax expression, and increased TUNEL-positive apoptotic nuclei characterized the restored epithelia of FHIT transduced forestomachs. Thus, FHIT viral gene delivery prevents or retards development of carcinogen-induced forestomach tumors and reverses development of established tumors by 60-70% through an apoptotic pathway. This dramatic reduction in tumor burden emphasizes the efficacy of targeting the FHIT apoptotic pathway for tumor eradication.


Assuntos
Hidrolases Anidrido Ácido , Dimetilnitrosamina/análogos & derivados , Proteínas de Neoplasias/genética , Neoplasias Gástricas/terapia , Administração Oral , Animais , Apoptose , Divisão Celular , Mucosa Gástrica/patologia , Terapia Genética , Vetores Genéticos/administração & dosagem , Camundongos , Modelos Biológicos , Estômago/anatomia & histologia , Estômago/patologia , Neoplasias Gástricas/induzido quimicamente , Neoplasias Gástricas/patologia , Vírus/genética
16.
Clin Cancer Res ; 10(22): 7607-12, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15569992

RESUMO

The fragile histidine triad (FHIT) gene located on chromosome 3p14.2 is frequently deleted in human tumors. We have previously reported deletions at the FHIT locus in 50% of bladder carcinoma derived cell lines and reduced expression in 61% of primary transitional carcinomas of the urinary bladder. To additionally investigate the role of FHIT alterations in the development of bladder cancer, we used heterozygous and nullizygous Fhit-deficient mice in a chemically induced carcinogenesis model. Results showed that 8 of 28 (28%) and 6 of 13 (46%) of the Fhit -/- and +/-, respectively, versus 2 of 25 (8%) Fhit +/+ mice developed invasive carcinoma after treatment with N-butyl-N-(4-hydroxybutyl) nitrosamine. To explore the possibility of a FHIT-based gene therapy for bladder cancer, we studied the effects of restored Fhit protein expression on cell proliferation, cell kinetics, and tumorigenicity in BALB/c nude mice, with human SW780 Fhit-null transitional carcinoma derived cells. In vitro transduction of SW780 Fhit-negative cells with adenoviral-FHIT inhibited cell growth, increased apoptotic cell population, and suppressed s.c. tumor growth in nude mice. These findings suggest the important role of Fhit in bladder cancer development and support the effort to additionally investigate a FHIT-based gene therapy.


Assuntos
Hidrolases Anidrido Ácido/genética , Carcinoma de Células de Transição/genética , Proteínas de Neoplasias/genética , Neoplasias da Bexiga Urinária/genética , Adenoviridae/genética , Animais , Butilidroxibutilnitrosamina/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Terapia Genética/métodos , Genótipo , Heterozigoto , Humanos , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fatores de Tempo
17.
Curr Gene Ther ; 4(1): 53-63, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15032614

RESUMO

A link between common chromosome fragile sites and frequent chromosomal deletions in cancer was observed two decades ago and led to the hypothesis that genes at fragile sites may play a role in tumor development. In 1996, the human fragile histidine triad gene, FHIT, was identified by positional cloning of the chromosome region spanning the carcinogen-sensitive, common fragile site, FRA3B at 3p14.2. Loss or inactivation of the FHIT gene in a large fraction of human tumors results in absence or reduction of Fhit protein. In vitro analyses and in vivo tumorigenicity studies show that restoration of Fhit protein induces tumor suppression in 50% of tumor cell lines tested. Viral vector-mediated FHIT gene transfer to Fhit-deficient mice not only prevents but reverses the carcinogen-induced tumor development in vivo, in accordance with the oncosuppressive properties of Fhit protein. The strong proapoptotic activity following Fhit infection of cancer cells strengthens the case for further exploration of FHIT gene therapy in cancer prevention and treatment.


Assuntos
Hidrolases Anidrido Ácido/genética , Terapia Genética , Proteínas de Neoplasias/genética , Neoplasias/terapia , Hidrolases Anidrido Ácido/metabolismo , Animais , Apoptose , Carcinógenos Ambientais/toxicidade , Transformação Celular Neoplásica/genética , Sítios Frágeis do Cromossomo/genética , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos , Mutação , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/prevenção & controle
18.
Carcinogenesis ; 27(7): 1489-96, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16543248

RESUMO

Upper aerodigestive tract (UADT) cancer, including oral and esophageal cancer, is an important cause of cancer deaths worldwide. Patients with UADT cancer are frequently zinc deficient (ZD) and show a loss of function of the pivotal tumor suppressor gene p53. The present study examined whether zinc deficiency in collaboration with p53 insufficiency (p53+/-) promotes lingual and esophageal tumorigenesis in mice exposed to low doses of the carcinogen 4-nitroquinoline 1-oxide. In wild-type mice, ZD significantly increased the incidence of lingual and esophageal tumors from 0% in zinc sufficient (ZS) ZS:p53+/+ mice to approximately 40%. On the p53+/- background, ZD:p53+/- mice had significantly greater tumor incidence and multiplicity than ZS:p53+/- and ZD:p53+/+ mice, with a high frequency of progression to malignancy. Sixty-nine and 31% of ZD:p53+/- lingual and esophageal tumors, respectively, were squamous cell carcinoma versus 19 and 0% of ZS:p53+/- tumors (tongue, P = 0.003; esophagus, P = 0.005). Immunohistochemical analysis revealed that the increased cellular proliferation observed in preneoplastic lingual and esophageal lesions, as well as invasive carcinomas, was accompanied by overexpression of cytokeratin 14, cyclooxygenase-2 and metallothionein. In summary, a new UADT cancer model is developed in ZD:p53+/- mouse that recapitulates aspects of the human cancer and provides opportunities to probe the genetic changes intrinsic to UADT carcinogenesis and to test strategies for prevention and reversal of this deadly cancer.


Assuntos
Neoplasias Esofágicas/patologia , Neoplasias da Língua/patologia , Proteína Supressora de Tumor p53/deficiência , Zinco/deficiência , 4-Nitroquinolina-1-Óxido/toxicidade , Animais , Biomarcadores Tumorais/análise , Carcinógenos/toxicidade , Ciclo-Oxigenase 2/biossíntese , Modelos Animais de Doenças , Neoplasias Esofágicas/induzido quimicamente , Neoplasias Esofágicas/genética , Feminino , Expressão Gênica , Predisposição Genética para Doença , Imuno-Histoquímica , Queratina-14 , Queratinas/biossíntese , Masculino , Metalotioneína/biossíntese , Camundongos , Camundongos Mutantes , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Neoplasias da Língua/induzido quimicamente , Neoplasias da Língua/genética
19.
J Natl Cancer Inst ; 97(1): 40-50, 2005 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-15632379

RESUMO

BACKGROUND: Cancer of the upper aerodigestive tract, including esophageal and tongue carcinomas, is a major cause of cancer deaths worldwide. Esophageal and tongue cancers have both been associated with dietary zinc deficiency (ZD), and cyclooxygenase (COX-2) is often overexpressed in these cancers. Using rat models, we examined whether zinc regulates COX-2 expression in these cancers. METHOD: Expression of COX-2 protein and mRNA in rat lingual and esophageal epithelia in control (zinc sufficient [ZS]) rats, during ZD, and after intragastric zinc replenishment (ZR) were determined by immunoblotting, immunohistochemistry, and real-time quantitative polymerase chain reaction. COX-2 gene expression, cell proliferation, and apoptosis were analyzed in ZD, ZR, and ZD rats treated with the COX-2 inhibitors celecoxib and indomethacin. Tumor development in ZD rats treated by continuous exposure to the carcinogen 4-nitroquinoline 1 oxide (NQO), which causes tongue tumors in rats, was compared with those in NQO-treated ZS rats. Statistical tests were two-sided. RESULTS: The esophagus and tongue of ZD rats were hyperplastic and expressed COX-2 protein and mRNA at 8- to 14.7-fold higher levels than control rats. Within hours ZR reduced COX-2 overexpression to threefold that in control rats and reversed the hyperplastic phenotypes. The esophagus of ZD rats treated with celecoxib or indomethacin showed a reduction in cell proliferation and stimulation of apoptosis. NQO treatment resulted in greater incidence of lingual squamous cell carcinomas (74% versus 22%, difference = 52%, 95% confidence interval [CI] = 20% to 80%, P = .015) and greater tumor multiplicity (13.1 versus 4.3, difference = 8.8, 95% CI = 7.0 to 10.6, P = .018) in ZD than ZS rats. Of 23 NQO-treated ZD rats, 39% (9) and 61% (14) harbored esophageal and forestomach tumors, respectively, whereas none of the NQO-treated ZS rats did. CONCLUSIONS: COX-2 overexpression accompanies hyperplasia in ZD rats. Increased cell proliferation in NQO-treated ZD rats facilitates the development of tumors at multiple sites. The finding that zinc regulates COX-2 expression in vivo in an animal model may lead to prevention or therapeutic possibilities for upper aerodigestive tract cancer.


Assuntos
Dieta , Neoplasias Esofágicas/enzimologia , Isoenzimas/efeitos dos fármacos , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintases/efeitos dos fármacos , Prostaglandina-Endoperóxido Sintases/metabolismo , Neoplasias da Língua/enzimologia , Zinco/sangue , Zinco/farmacologia , 4-Nitroquinolina-1-Óxido , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Carcinógenos , Carcinoma de Células Escamosas/enzimologia , Celecoxib , Proliferação de Células/efeitos dos fármacos , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Modelos Animais de Doenças , Neoplasias Esofágicas/induzido quimicamente , Neoplasias Esofágicas/prevenção & controle , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hiperplasia/induzido quimicamente , Hiperplasia/prevenção & controle , Indometacina/farmacologia , Isoenzimas/genética , Masculino , Mutagênicos , Prostaglandina-Endoperóxido Sintases/genética , Pirazóis/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfonamidas/farmacologia , Neoplasias da Língua/induzido quimicamente , Neoplasias da Língua/prevenção & controle , Regulação para Cima/efeitos dos fármacos , Zinco/administração & dosagem , Zinco/deficiência
20.
Mol Carcinog ; 43(3): 175-81, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15937960

RESUMO

DNA hypermethylation is associated with decreased expression of tumor suppressor genes. We previously observed decreased Fhit expression and Fhit promoter region hypermethylation in rodent tumors induced by various carcinogens, and noted that the 5' regulatory regions in the promoter, exon 1, and intron 1 were differentially methylated, depending on the tissue of origin. Because different carcinogens were used for induction of tumors of the different organs, we could not conclude that the methylation patterns were tissue-specific. To determine if in rat tissues: (1) Fhit methylation status is related to expression levels and (2) Fhit methylation patterns were tissue- or carcinogen-specific, we examined Fhit methylation status and expression levels in DMBA- and MNU-induced benign and malignant mammary tumors. Fhit intron 1 was methylated in 3/9 DMBA and all of MNU-induced benign mammary tumors, in association with reduced Fhit expression levels; Fhit promoter and intron 1 were methylated in all DMBA and MNU-induced carcinomas in association with highly reduced Fhit expression levels. Treatment of rat cancer cells in vitro with the DNA methyltransferase inhibitor, 5'-Aza-2'deoxycytidine, for 4 d, increased Fhit expression and altered the methylation status. Before treatment, both promoter and intron 1 regions were methylated; after treatment, only intron 1 remained methylated. Thus, in carcinogen-exposed rat tissues there is an overall association of Fhit expression with regulatory region methylation, and hypermethylation patterns did not vary with carcinogen. The specific patterns of hypermethylated CpGs in the Fhit regulatory regions thus appear to be tissue-specific.


Assuntos
Hidrolases Anidrido Ácido/genética , Metilação de DNA , Regulação da Expressão Gênica , Genes Reguladores , Genes Supressores de Tumor , Proteínas de Neoplasias/genética , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Sequência de Bases , Carcinógenos/toxicidade , Metilação de DNA/efeitos dos fármacos , Primers do DNA , Feminino , Genes Reguladores/efeitos dos fármacos , Íntrons , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Metilnitrosoureia/toxicidade , Especificidade de Órgãos , Reação em Cadeia da Polimerase , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA